diarhea chemo drug induced ncbi

13
Chemotherapy-induced diarrhea: pathophysiology, frequency and guideline-based management Alexander Stein, Wieland Voigt and Karin Jordan Abstract: Diarrhea is one of the main drawbacks for cancer patients. Possible etiologies could be radiotherapy, chemotherapeutic agents, decreased physical performance, graft versus host disease and infections. Chemotherapy-induced diarrhea (CID) is a common problem, especially in patients with advanced cancer. The incidence of CID has been reported to be as high as 5080% of treated patients (30% CTC grade 35), especially with 5-fluorouracil bolus or some combination therapies of irinotecan and fluoropyrimidines (IFL, XELIRI). Regardless of the molecular targeted approach of tyrosine kinase inhibitors and antibodies, diarrhea is a common side effect in up to 60% of patients with up to 10% having severe diarrhea. Furthermore, the underlying pathophysiology is still under investigation. Despite the number of clinical trials evaluating therapeutic or prophylactic measures in CID, there are just three drugs recommended in current guidelines: loperamide, deodorized tincture of opium and octreotide. Newer strategies and more effective agents are being developed to reduce the morbidity and mortality associated with CID. Recent research focusing on the prophylactic use of antibiotics, budesonide, probiotics or activated charcoal still have to define the role of these drugs in the routine clinical setting. Whereas therapeutic management and clinical work-up of patients presenting with diarrhea after chemotherapy are rather well defined, prediction and prevention of CID is an evolving field. Current research focuses on establishing predictive factors for CID like uridine diphosphate glucuronosyltransferase-1A1 polymorphisms for iri- notecan or dihydropyrimidine-dehydrogenase insufficiency for fluoropyrimidines. Keywords: chemotherapy-induced diarrhea, frequency, irinotecan, loperamide, octreotide, pathophysiology, prevention management Introduction In oncological patients, diarrhea can occur in several different situations. Possible etiologies could be radiotherapy, chemotherapeutic agents, decreased physical performance, graft versus host disease and infections. Careful analy- sis of the causative agent can lead to a more accu- rate management and early intervention possibly helps to prevent severe complications that may be irreversible [Davila and Bresalier, 2008; Vincenzi et al. 2008]. In particular, chemotherapy-induced diarrhea (CID) is a common problem in patients with advanced cancer and has to be carefully dif- ferentiated from other causes of diarrhea [Gibson and Stringer, 2009]. Chemotherapy-induced diarrhea CID can occur in 5080% of patients depending on the chemotherapy regimen [Benson et al. 2004; Gibson and Stringer, 2009]. A review of early toxic deaths occurring in two National Cancer Institute-sponsored cooperative group trials of irinotecan plus high-dose fluorouracil and leucovorin for advanced colorectal cancer has led to the recognition of a life-threatening gastrointestinal syndrome and highlighted the need for vigilant monitoring and aggressive ther- apy for this serious complication [Conti et al. 1996; Arbuckle et al. 2000; Saltz et al. 2000]. CID can cause depletion of fluids and electro- lytes, malnutrition, dehydration and hospitaliza- tion, all of which can lead to cardiovascular http://tam.sagepub.com 51 Therapeutic Advances in Medical Oncology Review Ther Adv Med Oncol (2010) 2(1) 5163 DOI: 10.1177/ 1758834009355164 ! The Author(s), 2010. Reprints and permissions: http://www.sagepub.co.uk/ journalsPermissions.nav Correspondence to: Alexander Stein, MD Department of Internal Medicine IV, Oncology/ Hematology/ Hemostaseology, Martin-Luther-University Halle/Wittenberg, Ernst-Grube-Str. 40, 06120 Halle/Saale, Germany alexander.stein@ medizin.uni-halle.de Wieland Voigt Karin Jordan Department of Internal Medicine IV, Oncology/ Hematology/ Hemostaseology, Martin-Luther-University Halle/Wittenberg, Ernst-Grube-Str. 40, 06120 Halle/Saale, Germany

Upload: faris-azhar

Post on 16-Sep-2015

216 views

Category:

Documents


3 download

DESCRIPTION

diare

TRANSCRIPT

  • Chemotherapy-induced diarrhea:pathophysiology, frequency andguideline-based management

    Alexander Stein, Wieland Voigt and Karin Jordan

    Abstract: Diarrhea is one of the main drawbacks for cancer patients. Possible etiologies couldbe radiotherapy, chemotherapeutic agents, decreased physical performance, graft versus hostdisease and infections. Chemotherapy-induced diarrhea (CID) is a common problem, especiallyin patients with advanced cancer. The incidence of CID has been reported to be as high as5080% of treated patients (30% CTC grade 35), especially with 5-fluorouracil bolus orsome combination therapies of irinotecan and fluoropyrimidines (IFL, XELIRI). Regardless ofthe molecular targeted approach of tyrosine kinase inhibitors and antibodies, diarrhea is acommon side effect in up to 60% of patients with up to 10% having severe diarrhea.Furthermore, the underlying pathophysiology is still under investigation. Despite the number ofclinical trials evaluating therapeutic or prophylactic measures in CID, there are just threedrugs recommended in current guidelines: loperamide, deodorized tincture of opium andoctreotide. Newer strategies and more effective agents are being developed to reduce themorbidity and mortality associated with CID. Recent research focusing on the prophylactic useof antibiotics, budesonide, probiotics or activated charcoal still have to define the role of thesedrugs in the routine clinical setting. Whereas therapeutic management and clinical work-up ofpatients presenting with diarrhea after chemotherapy are rather well defined, prediction andprevention of CID is an evolving field. Current research focuses on establishing predictivefactors for CID like uridine diphosphate glucuronosyltransferase-1A1 polymorphisms for iri-notecan or dihydropyrimidine-dehydrogenase insufficiency for fluoropyrimidines.

    Keywords: chemotherapy-induced diarrhea, frequency, irinotecan, loperamide, octreotide,pathophysiology, prevention management

    IntroductionIn oncological patients, diarrhea can occur in

    several different situations. Possible etiologies

    could be radiotherapy, chemotherapeutic

    agents, decreased physical performance, graft

    versus host disease and infections. Careful analy-

    sis of the causative agent can lead to a more accu-

    rate management and early intervention possibly

    helps to prevent severe complications that may be

    irreversible [Davila and Bresalier, 2008; Vincenzi

    et al. 2008]. In particular, chemotherapy-induced

    diarrhea (CID) is a common problem in patients

    with advanced cancer and has to be carefully dif-

    ferentiated from other causes of diarrhea [Gibson

    and Stringer, 2009].

    Chemotherapy-induced diarrheaCID can occur in 5080% of patients dependingon the chemotherapy regimen [Benson et al.

    2004; Gibson and Stringer, 2009]. A review of

    early toxic deaths occurring in two National

    Cancer Institute-sponsored cooperative group

    trials of irinotecan plus high-dose fluorouracil

    and leucovorin for advanced colorectal cancer

    has led to the recognition of a life-threatening

    gastrointestinal syndrome and highlighted the

    need for vigilant monitoring and aggressive ther-

    apy for this serious complication [Conti et al.

    1996; Arbuckle et al. 2000; Saltz et al. 2000].

    CID can cause depletion of fluids and electro-

    lytes, malnutrition, dehydration and hospitaliza-

    tion, all of which can lead to cardiovascular

    http://tam.sagepub.com 51

    Therapeutic Advances in Medical Oncology Review

    Ther Adv Med Oncol

    (2010) 2(1) 5163

    DOI: 10.1177/1758834009355164

    ! The Author(s), 2010.Reprints and permissions:http://www.sagepub.co.uk/journalsPermissions.nav

    Correspondence to:Alexander Stein, MDDepartment of InternalMedicine IV, Oncology/Hematology/Hemostaseology,Martin-Luther-UniversityHalle/Wittenberg,Ernst-Grube-Str. 40,06120 Halle/Saale,[email protected]

    Wieland VoigtKarin JordanDepartment of InternalMedicine IV, Oncology/Hematology/Hemostaseology,Martin-Luther-UniversityHalle/Wittenberg,Ernst-Grube-Str. 40,06120 Halle/Saale,Germany

  • compromise and death. In addition, diarrhea can

    interfere with and detract from cancer treatment

    by causing dosing delays or reductions which

    may have an impact on survival [Engelking

    et al. 1998; Ippoliti, 1998]. Therapeutic agents

    commonly causing diarrhea include

    5-fluorouracil (5-FU), capecitabine and irinote-

    can (CPT-11) [Benson et al. 2004; Keefe et al.

    2004]. Usually it is a dose-related adverse effect

    and may be associated with other features of tox-

    icity. CID appears to be a multifactorial process

    whereby acute damage to the intestinal mucosa

    (including loss of intestinal epithelium, superfi-

    cial necrosis and inflammation of the bowel

    wall) causes an imbalance between absorption

    and secretion in the small bowel [Keefe et al.

    2000; Keefe, 2007; Gibson and Stringer, 2009].

    FrequencyThe frequency of CID depends on the drug and

    schedule, with the highest rate of diarrhea occur-

    ring with weekly irinotecan and bolus 5-FU

    (Table 1). Late diarrhea from irinotecan occurs

    at all dose levels, whereas early-onset diarrhea

    (24 hours after administration) is dose depen-dent, developing in up to 10% of patients (grade

    3/4). The median time to onset of late diarrhea is

    about 6 days with the 350mg/m2 every 3 weeks

    schedule and 11 days with the weekly schedule

    (125mg/m2).

    Fluoropyrimidines have also been associated with

    severe diarrhea. Both the therapeutic efficacy and

    frequency of diarrhea associated with 5-FU are

    increased when given with leucovorin (LV).

    Clinical manifestations and evaluationCID can be debilitating and, in some cases, life

    threatening. Findings in such patients include

    volume depletion, renal failure, and electrolyte

    disorders such as metabolic acidosis and depend-

    ing upon water intake, hyponatremia (increased

    water intake that cannot be excreted because of

    the hypovolemic stimulus to the release of anti-

    diuretic hormone) or hypernatremia (insufficient

    water intake to replace losses) [Benson et al.

    2004; Maroun et al. 2007].

    Diagnosis of CID begins with a history to deter-

    mine the severity according to the NCI CTC

    grades (recently updated National Cancer

    Institute Common Toxicity Criteria, Table 2).

    The volume and duration of diarrhea should

    also be determined, and the history should

    include questions concerning foods or drugs

    that might play a contributory role.

    It should also be considered that other factors

    can contribute to diarrhea in cancer patients trea-

    ted with 5-FU or irinotecan. These include intes-

    tinal infection (e.g. Clostridium difficile), radiation,

    and a history of prior intestinal resection [Davila

    and Bresalier, 2008; Vincenzi et al. 2008].

    Irinotecan-induced diarrheaIrinotecan is frequently used in first- and

    second-line treatment of metastatic colorectal

    cancer [Saltz et al. 2000, 2007; Hurwitz et al.

    2004; Jordan et al. 2004; Van Cutsem et al.

    2009]. Regardless of its schedule of

    Table 1. Rates of grade 3/4 diarrhea (CTC grades) for different therapeutic agents and combinations.

    Agent Grade 3/4 diarrhea

    Chemotherapy Single agent Combination therapy5-FU (bolus) 32% (G3) 26% XELIRI5-FU (CI) 613% 2528% IFL (bolus)irinotecan (late diarrhea) 1622% 1114% FOLFIRI (bolus/CI)capecitabine 11%docetaxel/paclitaxel 4% 14% docetaxel + capecitabine

    19% DCFTargeted agentsanti-EGFR-antibodies 12% 15% cetuximab +FOLFIRIanti-EGFR-TKI 69% 13% lapatinib + capecitabine

    15% lapatinib + paclitaxel6% erlotinib + gemcitabine

    sorafenib/sunitinib 28% (G3)m-TOR inhibitors 14% (G3)

    5-FU, 5-fluorouracil; DCF, docetaxel + cisplatin + 5-FU; EGFR, epidermal growth factor receptor; FOLFIRI,irinotecan + leucovorin + 5-FU; IFL, irinotecan + leucovorin + 5-FU; m-TOR, mammalian target of rapamycin; TKI, tyrosinekinase inhibitor; XELIRI, irinotecan + capecitabine.

    Therapeutic Advances in Medical Oncology 2 (1)

    52 http://tam.sagepub.com

  • administration, myelosuppression and delayed-

    type diarrhea are the most common side effects

    [Davila and Bresalier, 2008].

    Irinotecan can cause acute diarrhea (immediately

    after drug administration) or delayed diarrhea.

    Immediate-onset diarrhea is caused by acute cho-

    linergic properties and is often accompanied by

    other symptoms of cholinergic excess, including

    abdominal cramping, rhinitis, lacrimation, and

    salivation. The mean duration of symptoms is

    30 minutes and they usually respond rapidly to

    atropine. Delayed-type diarrhea is defined as

    diarrhea occurring more than 24 hours after

    administration of irinotecan and is noncumula-

    tive and occurs at all dose levels.

    Main clinical predictive factors for irinotecan-

    related diarrhea are weekly administration, poor

    performance status, high serum creatinine levels,

    prior abdominopelvic irradiation, low leukocyte

    counts, age over 70 years, Gilbert syndrome

    and Crigler-Najjar syndrome type 1 [Vincenzi

    et al. 2008].

    Pathophysiology of irinotecan-induced diarrheaIrinotecan is converted by hepatic and peripheral

    carboxylesterase to its active metabolite

    7-ethyl-10-hydroxycamptothecin (SN38), which

    is subsequently glucuronidated by hepatic uri-

    dine diphosphate glucuronosyltransferase-1A1

    (UDP-GT 1A1) to SN38-glucuronide (SN38G)

    as depicted in Figure 1 [Voigt et al. 1998; Gibson

    and Stringer, 2009].

    Both SN-38 and SN-38G are excreted via urine

    and bile. Mass balance studies using 14car-

    bonlabelled irinotecan have demonstrated thatthe fecal route of excretion is the major route

    eliminating 63.7% of the administered drug.

    SN38G, once in the intestinal lumen, is deconju-

    gated by bacterial -glucuronidase to SN38. In

    feces, SN38 was found to be in excess relative to

    SN38G, which is suggestive of substantial

    -glucuronidase activity in the human intestinal

    contents [Saliba et al. 1998; Stringer et al. 2008].

    The free intestinal luminal SN38, either from bile

    or SN38G deconjugation, is responsible for

    irinotecan-induced diarrhea. The different

    mechanisms in detail by which the free SN38

    induces diarrhea are still a matter of debate

    [Stringer et al. 2007].

    In summary the different mechanisms are dis-

    cussed as follows:

    (1) Free intestinal luminal SN38 induces directmucosal damage with water and electrolytemalabsorption and mucous hypersecretionin rats [Takasuna et al. 1995].

    (2) The luminal environment is alteredby irinotecan and as a result may favordifferent genera of bacteria, allowing them

    Table 2. Common Toxicity Criteria (version 3.0 and 4.02) for diarrhea, adapted from the National Cancer Institute.

    Grade

    1 2 3 4 5

    Diarrhea(version 3.0)

    Increase of

  • to proliferate. The bacterial -glucuronidasethen deconjugates SN38G to the activeform SN38 at an increased rate causing sig-nificant damage and diarrhea. [Takasunaet al. 1998; Stringer et al. 2007, 2008].

    (3) The distribution and severity of histologicaldamage within the rat intestine after admin-istration of irinotecan has been correlatedto the luminal -glucuronidase activity inrodents [Takasuna et al. 1998; Fittkauet al. 2004].

    (4) Irinotecan causes severe colonic damage(increased apoptosis, crypt hypoplasia anddilation) with accompanying excessivemucous secretion, as well as the usualchemotherapy-induced small intestinaldamage, like villous atrophy and crypt hypo-plasia. Increased levels of cell apoptosiscombined with the histopathologicalchanges in both the jejunum and colonand the changes in goblet cell numbersmay cause changes in absorption rates,possibly leading to diarrhea [Gibson et al.2003].

    (5) Irinotecan causes an increase in mucinsecretion, accompanied by a significantdecrease of mucin expression in the jejunumand colon of rats shown by immunohisto-chemistry for Muc2 and Muc4. Thereforethe increase of mucin secretion is likely tobe related to altered mucine gene expres-sion, and may contribute to diarrheainduced by irinotecan [Stringer et al. 2009].

    Molecular factors predictive ofirinotecan-induced toxicitiesConsidering the complex metabolism of irinote-

    can (Figure 1) there are a couple of molecular

    factors that are potentially predictive for

    toxicities. There is no established factor for the

    prediction of irinotecan-induced diarrhea yet.

    However, pharmacogenomic research revealed a

    predictive factor for hematologic toxicities, the

    UGT isoform, UDP-glucuronosyltransferase,

    or UGT1A1. UGT1A1 was one of the first fac-

    tors to be investigated, due to the observation of

    severe toxicities in patients with inherited disor-

    ders characterized by decreased bilirubin glucur-

    onidation like Gilberts syndrome (i.e. mild

    unconjugated hyperbilirubinemia) [Wasserman

    et al. 1997a]. Patients with homozygosity for

    UGT1A1*28 allele have lower UGT1A1 expres-

    sion, a decreased SN-38 glucuronidation and

    therefore a higher risk for developing severe iri-

    notecan toxicities. [Iyer et al. 2002; Innocenti

    et al. 2004]. Regarding a frequency of around

    9% for the homozygous allele, every tenth patient

    has an enhanced risk for hematological toxicities,

    leading to the approval of a genotyping method

    by the US Food and Drug Administration in

    2005. [Hoskins et al. 2007; Kim et al. 2007].

    A recommendation for an upfront dose reduction

    in this group of patients was added to the irino-

    tecan package insert. However, recent studies

    reveal varying results regarding the need for

    Hepatic cell membrane

    Irinotecan

    Irinotecan

    SN-38 SN-38G

    M4, APC, NPC

    SN-38G

    SN-38

    CES1CES2

    SLC01B1 ABCC1

    CYP3A4 CYP3A5

    UGT1A1 UGT1A9

    Via blood

    ABCB1 ABCC2 ABCG2

    ABCB1 ABCC2

    Bacterial -glucuronidase

    UGT1A1 UGT1A10

    SN-38

    Via bile excretedto the intestine

    Figure 1. Metabolism of irinotecan. UGT, UDP glucuronosyltransferase; SN-38, 7-ethyl-10-hydroxycamp-tothecin;, CYP, cytochrome P450;, CES carboxylesterases; APC, 7-ethyl-10-[4-N-(5-aminopentanoic acid)-1-piperidino]carbonyloxycamptothecin; NPC, 7-ethyl-10-(4-amino-1-piperidino)carbonyloxy-camptothecin; M,oxidized metabolite; ABCB/C, ATP-binding cassette, sub-family B/C.

    Therapeutic Advances in Medical Oncology 2 (1)

    54 http://tam.sagepub.com

  • dose reductions during irinotecan treatment in

    case of UGT1A1*28 genotype. In a retrospective

    analysis of the Dutch CAIRO trial, a reduced

    performance status and not the UGT1A1*28

    genotype was a predictor for febrile neutropenia

    in a bivariate analysis [Kweekel et al. 2008;

    Liu et al. 2008].

    Recently, the role of other UGT1A1 polymorph-

    isms and genetic variations of SLCO1B1 and

    ABC-transporters were investigated, with the

    latter playing a pivotal role in the excretion of

    SN-38 in the active form into the blood and in

    the glucuronidated form into the bile (Figure 1)

    [De Jong et al. 2007; Han et al. 2009; Innocenti

    et al. 2009]. The variability of the ABCC2 gene

    seems to be a determinant for irinotecan induced

    diarrhea. However, the clinical relevance of these

    factors still has to be determined. Further

    research to establish predictive factors for daily

    practice is absolutely essential.

    Fluoropyrimidines (5-FU, capecitabine,tegafur/uracil)The severity and prevalence of diarrhea caused

    by 5-FU treatment is increased by the addition

    of leucovorin (LV) to the treatment regimen.

    Diarrhea is reported in up to 50% of patients

    receiving weekly 5-FU/LV combined treatment.

    Moreover, the severity of the diarrhea can

    increase when 5-FU is administered by bolus

    injection as opposed to intravenous infusion

    [Vincenzi et al. 2008]. Clinical factors predictive

    for fluoropyrimidine-induced diarrhea are female

    sex, caucasian race and presence of diabetes

    [Zalcberg et al. 1998; McCollum et al. 2002;

    Meyerhardt et al. 2004]. The gender- and

    race-related differences are possibly influenced

    by the variable activity of dihydropyrimidine-

    dehydrogenase (DPD) [Mattison et al. 2006a].

    The leading polymorphism, which accounts for

    nearly 50% of nonfunctional alleles, is the

    DPYD*2A, resulting in a decreased drug clear-

    ance and prolonged exposure with severe toxici-

    ties. Complete DPD deficiency is extremely rare,

    but a partial deficiency is present in 35% of allcancer patients. DPD activity can be evaluated by

    peripheral blood mononuclear cell radioassay,

    DPD radioassaygenotyping of DPYD gene by

    denaturing high performance liquid chromatog-

    raphy (DHPLC), or 2-13C uracil breath test

    (UraBT). The current genotyping strategies are

    not yet available for routine use [Yen and

    McLeod, 2007]. Potentially, the simple breath

    test (UraBT) could be used as a screening tool

    [Mattison et al. 2006b].

    Of further predictive value are polymorphisms of

    the thymidilate synthase (TS) and methylenete-

    trahydrofolate reductase (MTHFR) genes.

    However, taking into account the multifactorial

    nature of fluoropyrimidine induced diarrhea, in

    daily practice genotyping for DPD will be

    initiated after occurrence of unusual toxicity.

    Pathophysiology of fluoropyrimidine-induceddiarrheaAlthough 5-FU is routinely used in the treatment

    of cancer and is known to cause diarrhea, very

    few basic research papers have attempted to elu-

    cidate the mechanisms underlying the pathophy-

    siology. Early investigations revealed 5-FU being

    the causative agent for mitotic arrest of intestinal

    crypt cells, decrease of the relative fraction of vil-

    lous enterocytes and the surface area for resorp-

    tion [Siber et al. 1980]. Further research focused

    on different dose schedules of this cytotoxic agent

    using 5-FU in animal models [Cao et al. 1998].

    Incidence of diarrhea with molecularlytargeted agents

    Epidermal growth factor receptor-targetedtherapiesThe rate of severe diarrhea (grade 3/4) with epi-

    dermal growth factor receptor (EGFR) targeting

    therapies is less than 10%. For monoclonal anti-

    bodies (mAb), such as the chimeric IgG1 mAb

    cetuximab or the fully human IgG2 mAb panitu-

    mumab, rates of grade 2 diarrhea are up to 21%

    and for grade 3 (ie greater than 7 stools per day

    or requiring intravenous fluids) between 1 and

    2% [Van Cutsem et al. 2007; Davila and

    Bresalier, 2008; Vincenzi et al. 2008]. Diarrhea

    is more common in patients receiving small mol-

    ecule EGFR tyrosine kinase inhibitors (TKIs),

    such as erlotinib, gefitinib or lapatinib.

    Occurrence of diarrhea is up to 60% for all

    grades. Grade 3 diarrhea develops in about

    69%. However, dose reduction due toEGFR-targeting therapy induced diarrhea is

    seldom necessary. In combination with radiother-

    apy diarrhea could be a more serious problem for

    EGFR-targeting drugs.

    Multitargeting tyrosine kinase inhibitorsSorafenib and sunitinib cause diarrhea in

    3050% of patients (all grades) with a rate ofless than 10% of grade 3 diarrhea [Llovet et al.

    A Stein, W Voigt et al.

    http://tam.sagepub.com 55

  • 2008; Gore et al. 2009; Motzer et al. 2009].

    Imatinib, an inhibitor of the Bcr-Abl protein tyr-

    osine kinase, causes diarrhea in about 30% of the

    patients, but severe diarrhea is also rare.

    m-TOR inhibitorsEverolimus and temsirolimus (inhibitors of the

    mammalian target of rapamycin [m-TOR])

    were both recently approved for treatment of

    renal cell cancer, causing diarrhea in up to 40%

    with a rate of severe diarrhea in less than 5% of

    patients [Hudes et al. 2007; Motzer et al. 2008;

    Hess et al. 2009].

    Pathophysiology of molecularly targetedagent-induced diarrheaThe mechanisms of targeted agent-induced diar-

    rhea are not adequately investigated yet. The

    antitumor activity is based on apoptosis induc-

    tion, antiangiogenesis and tyrosine kinase inhibi-

    tion by targeting receptors or signaling pathways

    that are present in normal cells as well, including

    the mucosa. Increased levels of EGFR are found

    in inflamed mucosa, particulary in goblet cells,

    which seem to play a role in CID [Threadgill

    et al. 1995]. However, there was no increase in

    toxicity of head and neck radiation by addition of

    cetuximab in a phase III trial despite a possible

    correlation between EGFR targeting and matu-

    ration of squamous eptithelium of the tongue and

    nasal cavity [Bonner et al. 2006; Keefe and

    Gibson, 2007]. The high expression of Kit in

    the interstitial cells of Cajal, which function as

    pacemaker cells of the intestinal motility, might

    be a potential mechanism for diarrhea induced by

    imatinib or sunitinib [Deininger et al. 2003].

    Regarding the increasing utilization of targeted

    therapies further research to gain the ability to

    prevent diarrhea is urgently warranted [Keefe

    and Anthony, 2008].

    Therapeutic approachesThe treatment of CID includes nonpharmacolo-

    gic and pharmacologic interventions to control

    diarrhea and careful serial evaluation to rule out

    significant volume depletion or comorbidities

    that would require specific intervention or hospi-

    talization [Benson et al. 2004; Maroun et al.

    2007]. Initial nonpharmacologic measures

    include avoidance of foods that would aggravate

    the diarrhea and aggressive oral rehydration with

    fluids that contain water, salt, and sugar

    [Dupont, 1997]. These principles are similar to

    those used for infectious diarrhea.

    Given the lack of predictability for CID, signifi-

    cant effort has been made to evaluate prophylac-

    tic and therapeutic measures to reduce its

    severity. A broad variety of drugs have been

    tested for those measures.

    Prophylactic measures

    Antibiotics. Based on the assumption that bacte-rial -glucuronidase in the intestine is essential

    for activating SN-38G which plays a crucial

    role in the development of irinotecan-induced

    mucosal damage, the eradication of bacteria

    with marginally absorbable antibiotics, like neo-

    mycin, seems to be an interesting approach

    [Kehrer et al. 2001]. Despite promising results

    in a small series for secondary prophylaxis

    [Schmittel et al. 2004], a recent randomized

    phase II study displayed only a nonsignificant

    reduction of grade 3 diarrhea from 32.4 to

    17.9% [De Jong et al. 2006]. In contrast, in a

    further nonrandomized study with 51 patients

    using levofloxacin just one patient experienced

    grade 3 diarrhea with no grade 4 at all [Flieger

    et al. 2007]. Regarding these controversial

    results, the role of antibiotics in prevention of

    irinotecan-induced diarrhea has to be further

    investigated.

    Budesonide. Pathophysiologically, the reductionof inflammation in the bowel could possibly

    reduce the occurrence of diarrhea. The published

    data however, only reveal a trend towards a reduc-

    tion of CID from 4.2 to 1.8 days in a randomized

    phase II study when concomitantly loperamide

    was used [Karthaus et al. 2005]. In contrast,

    in loperamide-refractory patients the CID grade

    could be reduced in more than half of the

    patients treated with either irinotecan or 5-FU

    in a small case series [Lenfers et al. 1999].

    Larger studies are necessary to determine the

    actual role of budesonide.

    Glutamine. Preclinical data suggests that gluta-mine stimulates intestinal mucosa growth, dis-

    playing less gastrointestinal toxictiy in rodents

    treated with chemotherapy [Fox et al. 1988;

    Xue et al. 2008]. Results from randomized stu-

    dies showed a nonsignificant reduction in the

    CID rate [Daniele et al. 2001], and no effect in

    the prevention of radiation-induced diarrhea for

    the oral application form of glutamine was

    revealed [Kozelsky et al. 2003]. Importantly,

    a trial in patients receiving high-dose chemo-

    therapy and glutamine-containing intravenous

    Therapeutic Advances in Medical Oncology 2 (1)

    56 http://tam.sagepub.com

  • solutions showed significantly more relapses and

    deaths in the glutamine group [Pytlik et al. 2002].

    Recently, a series with 44 patients showed a sig-

    nificant reduction in diarrhea with prophylactic

    use of intravenous glutamine any influence

    on survival was not reported [Li et al. 2009].

    Considering these results, a further development

    of glutamine for CID seems to be questionable.

    Celecoxib. In animal models, celecoxibenhanced the antitumor activity of irinotecan

    and reduced the rate of diarrhea [Trifan et al.

    2002]. The rate of grade 3 diarrhea was only

    8% in one trial of 43 patients suffering from

    malignant gliomas treated with irinotecan and

    celecoxib [Reardon et al. 2005], whereas in

    another study with the same sample size using a

    combination of celecoxib and glutamine with

    the IFL-regimen the rate was 45% for grade 3

    diarrhea, which is even higher than the expected

    margin [Pan et al. 2005]. In a recent review, no

    improvement with the usage of celecoxib in redu-

    cing CID was observed in the analyzed studies

    [Fakih and Rustum, 2009].

    Long-acting formulation of octreotide. The effi-cacy of long-acting octreotide in the therapeutic

    setting has been demonstrated, as has its use in

    secondary prophylaxis in a small case series with

    doses ranging from 20mg up to 40mg every

    4 weeks [Rosenoff, 2004a,b]. A large randomized

    study resulted in a nonsignificant reduction of

    severe diarrhea (61.7 versus 48.4%) favoring a

    dose of 40mg over 30mg every 4 weeks as

    secondary prophylaxis [Rosenoff et al. 2006].

    However, preliminary results of a study presented

    by Zacchariah and colleagues at ASCO 2007 in

    the primary prophylaxis of diarrhea in 215 rectal

    cancer patients receiving 5-FU based chemora-

    diation was negative, revealing no difference

    between placebo and 30mg of long-acting

    octreotide [Zachariah et al. 2007]. In patients

    receiving pelvic radiation the prophylactic treat-

    ment with 20mg of long-acting octreotide versus

    placebo showed even worse tolerability regarding

    gastrointestinal symptoms and no change in diar-

    rhea [Martenson et al. 2008].

    Probiotics. Probiotics have been shown to pre-vent diarrhea in inflammatory bowel disease.

    Preclinical data yielded a similar efficacy in

    CID [Von Bultzingslowen et al. 2003; Bowen

    et al. 2007]. In the clinical setting, a combination

    of Lactobacillus rhamnosus and fiber resulted in

    a significant reduction of grade 3/4 diarrhea

    (37 versus 22%) in a randomized study of patients

    treated with either bolus (Mayo) or bolus and

    infusional (simplified de Gramont) 5-FU with

    leucovorin for adjuvant treatment of colorectal

    cancer [Osterlund et al. 2007].

    Activated charcoal. The prophylactic use of acti-vated charcoal in irinotecan-induced diarrhea

    seems to have interesting potential. Two small

    studies, one conducted in children, displayed a

    reduction in grade 3/4 diarrhea (7.1 versus 25%

    and 4.4 versus 52.3%) with excellent compliance

    and tolerability. The discontinuation rate of iri-

    notecan was much lower and less loperamide was

    used [Michael et al. 2004; Sergio et al. 2008].

    This approach should be further investigated in

    a phase III trial.

    A further possible approach is the modulation of

    irinotecan pharmacokinetics, by the addition

    of phenobarbital, phenytoin and cyclosporine,

    to downsize SN-38 biliary excretion and induce

    glucuronidation, limited by the small therapeutic

    range of the used drugs and the possible decre-

    mental impact on efficacy, due to reduced con-

    centration of active metabolites. Attempts have

    also been made to pharmacologically upregulate

    intestinal mucosal UDP-GT 1A1 with the plant

    flavonoid, chrysin. Other therapeutic measures

    assessed include an encephalinase inhibitor (acet-

    orphan), which seems to be equally effective

    as loperamide in the treatment of non-CID

    diarrhea.

    Guideline-based drug recommendationsSo far, only loperamide, octreotide and tincture

    of opium are recommended in the updated treat-

    ment guidelines by the consensus conference on

    the management of CID from Benson and col-

    leagues due to a lack of efficacy or insufficient

    evidence level of the other mentioned therapeutic

    approaches [Benson et al. 2004].

    OpioidsLoperamide is an opioid which functions by

    decreasing intestinal motility by directly affecting

    the smooth muscle of the intestine and has no

    systemic effects due to a minimal absorption.

    The recommendation in current treatment

    guidelines [Benson et al. 2004] is based on an

    effective reduction in fecal incontinence, fre-

    quency of bowel movements and stool weight.

    The dosage of loperamide is an initial 4mg

    dose followed by 2mg every 24 hours or after

    A Stein, W Voigt et al.

    http://tam.sagepub.com 57

  • every unformed stool. In case of CID, especially

    irinotecan-containing therapies, the more aggres-

    sive regimen should be chosen.

    Deodorized tincture of opium (DTO) is another

    widely used antidiarrheal agent, despite the

    absence of literature to support its use in CID

    treatment. DTO contains the equivalent of

    10mg/ml morphine. The recommended dose is

    1015 drops in water every 34 hours [Bensonet al. 2004]. The camphorated (alcohol-based)

    tincture is a less concentrated preparation con-

    taining the equivalent of 0.4mg/ml morphine,

    leading to a dose of 5ml (one teaspoon) every

    34 hours.

    OctreotideOctreotide, a synthetic somatostatin analog, acts

    via several mechanisms: decreased secretion of a

    number of hormones, such as vasoactive intesti-

    nal peptide (VIP); prolongation of intestinal tran-

    sit time and reduced secretion and increased

    absorption of fluid and electrolytes. It is approved

    by the US Food and Drug Administration for the

    treatment of diarrhea related to VIP-secreting

    tumors and symptoms due to carcinoid syn-

    drome. Octreotide is beneficial in patients with

    CID from fluoropyrimidines, irinotecan, and

    5-FU-based chemoradiotherapy [Gebbia et al.

    1993; Goumas et al. 1998; Barbounis et al.

    2001]. Although one randomized trial in 41

    5-FU-treated patients showed that octreotide

    was more effective than standard-dose lopera-

    mide (90 versus 15% resolution of diarrhea by

    day 3) [Cascinu et al. 1993], octreotide is gener-

    ally reserved as a second-line treatment for

    patients who are refractory after 48 hours, despite

    a loperamide escalation, because of its high cost

    [Zidan et al. 2001]. Patients developing a gastro-

    intestinal syndrome including severe diarrhea,

    nausea, vomiting, anorexia, and abdominal

    cramping should receive an aggressive manage-

    ment with intravenous fluids and upfront octreo-

    tide. These recommendations by the consensus

    conference mentioned above reflect the risk of

    life-threatening complications and the reduced

    activity of loperamide in cases of severe diarrhea

    [Cascinu et al. 2000].

    The optimal dosage of octreotide is not well

    defined. Current treatment guidelines recom-

    mend a starting dose of 100150mg subcuta-neously (sc) or intravenously (iv) three times

    a day. Doses could be escalated to 500 mg sc/ivthree times a day or by continuous iv infusion

    2550mg/hr showing a dose-response relation-ship without significant toxicities [Wadler et al.

    1995; Wasserman et al. 1997b].

    Summary of the consensus recommendationsThe recommendations of a consensus conference

    on the management of CID were published

    in 1998 and updated in 2004. Guidelines for

    evaluation and management of patients with

    CID are presented in Figure 2 [Wadler et al.

    1998, Benson et al. 2004]. The tempo and

    specific nature of treatment is guided by the

    classification of the symptom constellation as

    complicated or uncomplicated. Uncomplicated

    patients may be managed conservatively in the

    outpatient setting (at least initially), while those

    with severe diarrhea or a potentially exacerbating

    condition (eg abdominal cramping, nausea,

    vomiting, fever, sepsis, neutropenia or bleeding)

    should be admitted to the hospital and treated

    aggressively with octreotide, intravenous fluids,

    antibiotics and a diagnostic workup.

    ConclusionCID is caused by changes in intestinal absorption

    and might be accompanied by excessive electro-

    lyte and fluid secretion. Furthermore, this type of

    diarrhea may be a consequence of biochemical

    changes caused by chemotherapy. Depending

    on the chemotherapeutic regimen, rates of

    severe or life-threatening CID can be up to

    30% (grade 35 diarrhea), especially with 5-FUbolus or combination therapies of irinotecan and

    fluoropyrimidines (IFL, XELIRI). Regarding the

    tremendous effects on patients safety and quality

    of life, the possible occurrence of CID has to be

    carefully considered. Current research focuses

    on establishing predictive factors for toxicities

    caused by therapeutic agents like UGT1A1-

    polymorphisms for irinotecan or DPD-

    insufficiency for fluoropyrimidines. Despite the

    amount of clinical trials evaluating therapeutic

    or prophylactic measures in CID, there are just

    three drugs recommended in current guidelines:

    loperamide, deodorized tincture of opium and

    octreotide. Further evaluation of treatment

    options is absolutely essential for the manage-

    ment of this debilitating toxicity.

    Therapeutic Advances in Medical Oncology 2 (1)

    58 http://tam.sagepub.com

  • EVALUATE

    ADDED RISK FACTORS

    MANAGEMENT

    TREATMENT

    Reassess 1224 hours later Diarrhea unresolvedProgression to severe diarrhea(NCI grades 34 with or withoutfever, dehydration, neutropenia,and/or blood in stool)

    COMPLICATEDCTC grade 3 or 4 diarrhea

    Obtain history of onset and duration of diarrhea

    Assess patient for fever, dizziness, abdominal pain/cramping, or weakness (ie, rule out risk for sepsis, bowel obstruction, dehydration) Dietary profile (ie, to identify diarrhea-enhancing foods) Medications profile (ie, to identify diarrheogenic agents)

    Describe number of stools and stool composition (eg, watery, blood in stool, nocturnal)

    UNCOMPLICATEDCTC grade 1-2 diarrhea

    with no complicatingsigns or symptoms

    Cramping Nausea/vomiting ( grade 2) Decreased perofrmance stauts Fever Sepsis Neutropenia Frank bleeding Dehydration

    or grade 1 or 2 with one ormore of the following signsor symtoms

    Stop all lactose-containing products, alcohol, and high-osmolar supplements Drink 810 large glasses of clear liquids a day (eg, Gatorade or broth) Eat frequent small meals (eg, bananas, rice, appleasuce, toast, plain pasta) Instruct patient to record the number of stools and report symptoms of life-threatening sequelae (eg, fever or dizziness upon standing) For grade 2 diarrhea, hold cytotoxic chemotherapy unitl symptoms resolve and consider dose reduction

    Administer standard dose of loperamide: initial dose 4 mg followed by 2 mg every 4 hours or after every unformed stool Consider clinical trial

    Diarrhea resolving

    Diarrhea resolving

    Cotinue instructions for dietary modification

    Cotinue instructions for dietary modification

    Gradually add solid foods to diet

    Gradually add solid foods to diet

    Discontinue loperamide after 12-hour diarrhea-free interval

    Discontinue loperamide after 12-hour diarrhea-free interval

    RT-induced: continue loperamide

    RT-induced: continue loperamide

    Persistent diarrhea (NCI grades 12) Administer loperamide 2 mg every 2 hours Start oral antibiotics Observe patient for responseRT-induced: oral antibiotics not

    generally recommended

    Reassess 1224 hours later

    Progression to severe diarrhea(NCI grades 34 with or withoutfever, dehydration, neutropenia,and/or blood in stool)

    Diarrhea unresolved

    ADMIT TO HOSPITAL

    EVALUATE IN OFFICE/OUTPATIENT CENTER

    Persistent diarrhea (NCI grades 12)(no fever, dehydration, neutropenia,and/or blood in stool)

    Check stool workup(blood, fecal leukocytes, Clostridium difficile, Salmonella, Escherichia coli,Campylobacter, infectious colitis)

    Check CBC and electrolytes Perform abdominal exam Replace fluids and electrolytes as appropriate Discontinue loperamide and begin second-line agent

    Octreotide (100 to 150 g SC TID with dose escalation up to 500 g TID) Other second-line agent (eg, tincture of opium)

    RT-induced: Continue loperamide or other oral agent; no workup required

    Administer octreotide(100 to 150 g SC TID or IV (25-50 g/hr)if dehydration is severe with dose

    as needed (eg, fluorogquinolone)

    all symptoms resolve; restartchemotherapy at reduced dose

    escalation up to 500 g TID) Start intravenous fluids and antibiotics

    Stool work-up, CBC, and electrolyte profile Discontinue cytotoxic chemotherapy until

    Figure 2. Consensus guideline for the treatment of chemotherapy induced diarrhea [Benson et al. 2004].Reprinted with permission ! 2008 American Society of Clinical Oncology. All rights reserved.

    A Stein, W Voigt et al.

    http://tam.sagepub.com 59

  • Conflict of interest statementNone declared.

    ReferencesArbuckle, R.B., Huber, S.L. and Zacker, C. (2000)The consequences of diarrhea occurring during che-motherapy for colorectal cancer: a retrospective study.Oncologist 5: 250259.

    Barbounis, V., Koumakis, G., Vassilomanolakis, M.,Demiri, M. and Efremidis, A.P. (2001) Control ofirinotecan-induced diarrhea by octreotide after loper-amide failure. Support Care Cancer 9: 258260.

    Benson 3rd, A.B., Ajani, J.A., Catalano, R.B.,Engelking, C., Kornblau, S.M., Martenson Jr, J.A.et al. (2004) Recommended guidelines for the treat-ment of cancer treatment-induced diarrhea. J ClinOncol 22: 29182926.

    Bonner, J.A., Harari, P.M., Giralt, J., Azarnia, N.,Shin, D.M., Cohen, R.B. et al. (2006) Radiotherapyplus cetuximab for squamous-cell carcinoma of thehead and neck. N Engl J Med 354: 567578.

    Bowen, J.M., Stringer, A.M., Gibson, R.J., Yeoh, A.S.,Hannam, S. and Keefe, D.M. (2007) Vsl#3 probiotictreatment reduces chemotherapy-induced diarrheaand weight loss. Cancer Biol Ther 6: 14491454.

    Cao, S., Troutt, A.B. and Rustum, Y.M. (1998)Interleukin 15 protects against toxicity and potentiatesantitumor activity of 5-fluorouracil alone and in com-bination with leucovorin in rats bearing colorectalcancer. Cancer Res 58: 16951699.

    Cascinu, S., Fedeli, A., Fedeli, S.L. and Catalano, G.(1993) Octreotide versus loperamide in the treatmentof fluorouracil-induced diarrhea: a randomized trial. JClin Oncol 11: 148151.

    Cascinu, S., Bichisao, E., Amadori, D., Silingardi, V.,Giordani, P., Sansoni, E. et al. (2000) High-doseloperamide in the treatment of 5-fluorouracil-induceddiarrhea in colorectal cancer patients. Support CareCancer 8: 6567.

    Conti, J.A., Kemeny, N.E., Saltz, L.B., Huang, Y.,Tong, W.P., Chou, T.C. et al. (1996) Irinotecan is anactive agent in untreated patients with metastatic col-orectal cancer. J Clin Oncol 14: 709715.

    Daniele, B., Perrone, F., Gallo, C., Pignata, S., DeMartino, S., De Vivo, R. et al. (2001) Oral Glutaminein the prevention of fluorouracil induced intestinaltoxicity: a double blind, placebo controlled, rando-mised trial. Gut 48: 2833.

    Davila, M. and Bresalier, R.S. (2008) Gastrointestinalcomplications of oncologic therapy. Nat Clin PractGastroenterol Hepatol 5: 682696.

    De Jong, F.A., Kehrer, D.F., Mathijssen, R.H.,Creemers, G.J., De Bruijn, P., Van Schaik, R.H. et al.(2006) Prophylaxis of irinotecan-induced diarrheawith neomycin and potential role for Ugt1a1*28

    genotype screening: a double-blind, randomized,placebo-controlled study. Oncologist 11: 944954.

    De Jong, F.A., Scott-Horton, T.J., Kroetz, D.L.,McLeod, H.L., Friberg, L.E., Mathijssen, R.H. et al.(2007) Irinotecan-induced diarrhea: functional signif-icance of the polymorphic abcc2 transporter protein.Clin Pharmacol Ther 81: 4249.

    Deininger, M.W., OBrien, S.G., Ford, J.M. andDruker, B.J. (2003) Practical management of patientswith chronic myeloid leukemia receiving imatinib. JClin Oncol 21: 16371647.

    Dupont, H.L. (1997) Guidelines on acute infectiousdiarrhea in adults. The Practice ParametersCommittee of the American College ofGastroenterology. Am J Gastroenterol 92: 19621975.

    Engelking, C., Rutledge, D.N., Ippoliti, C., Neumann,J. and Hogan, C.M. (1998) Cancer-related diarrhea: aneglected cause of cancer-related symptom distress.Oncol Nurs Forum 25: 859860.

    Fakih, M.G. and Rustum, Y.M. (2009) Does celecoxibhave a role in the treatment of patients with colorectalcancer? Clin Colorectal Cancer 8: 1114.

    Fittkau, M., Voigt, W., Holzhausen, H.J. and Schmoll,H.J. (2004) Saccharic acid 1.4-lactone protects againstCpt-11-induced mucosa damage in rats. J Cancer ResClin Oncol 130: 388394.

    Flieger, D., Klassert, C., Hainke, S., Keller, R.,Kleinschmidt, R. and Fischbach, W. (2007) Phase IIclinical trial for prevention of delayed diarrhea withcholestyramine/levofloxacin in the second-line treat-ment with irinotecan biweekly in patients with meta-static colorectal carcinoma. Oncology 72: 1016.

    Fox, A.D., Kripke, S.A., De Paula, J., Berman, J.M.,Settle, R.G. and Rombeau, J.L. (1988) Effect of aglutamine-supplemented enteral diet onmethotrexate-induced enterocolitis. JPEN J ParenterEnteral Nutr 12: 325331.

    Gebbia, V., Carreca, I., Testa, A., Valenza, R., Curto,G., Cannata, G. et al. (1993) Subcutaneous octreotideversus oral loperamide in the treatment of diarrheafollowing chemotherapy. Anticancer Drugs 4: 443445.

    Gibson, R.J. and Stringer, A.M. (2009)Chemotherapy-induced diarrhoea. Curr Opin SupportPalliat Care 3: 3135.

    Gibson, R.J., Bowen, J.M., Inglis, M.R., Cummins,A.G. and Keefe, D.M. (2003) Irinotecan causes severesmall intestinal damage, as well as colonic damage, inthe rat with implanted breast cancer. J GastroenterolHepatol 18: 10951100.

    Gore, M.E., Szczylik, C., Porta, C., Bracarda, S.,Bjarnason, G.A., Oudard, S. et al. (2009) Safety andefficacy of sunitinib for metastatic renal-cell carci-noma: an expanded-access trial. Lancet Oncol10: 757763.

    Goumas, P., Naxakis, S., Christopoulou, A.,Chrysanthopoulos, C., Nikolopoulou, V.V. andKalofonos, H.P. (1998) Octreotide acetate in the

    Therapeutic Advances in Medical Oncology 2 (1)

    60 http://tam.sagepub.com

  • treatment of fluorouracil-induced diarrhea. Oncologist3: 5053.

    Han, J.Y., Lim, H.S., Park, Y.H., Lee, S.Y. and Lee,J.S. (2009) Integrated pharmacogenetic prediction ofirinotecan pharmacokinetics and toxicity in patientswith advanced non-small cell lung cancer. Lung Cancer63: 115120.

    Hess, G., Herbrecht, R., Romaguera, J., Verhoef, G.,Crump, M., Gisselbrecht, C. et al. (2009) Phase IIIstudy to evaluate temsirolimus compared with investi-gators choice therapy for the treatment of relapsed orrefractory mantle cell lymphoma. J Clin Oncol27: 38223829.

    Hoskins, J.M., Goldberg, R.M., Qu, P., Ibrahim, J.G.and McLeod, H.L. (2007) Ugt1a1*28 genotype andirinotecan-induced neutropenia: dose matters. J NatlCancer Inst 99: 12901295.

    Hudes, G., Carducci, M., Tomczak, P., Dutcher, J.,Figlin, R., Kapoor, A. et al. (2007) Temsirolimus,interferon alfa, or both for advanced renal-cellcarcinoma. N Engl J Med 356: 22712281.

    Hurwitz, H., Fehrenbacher, L., Novotny, W.,Cartwright, T., Hainsworth, J., Heim, W. et al. (2004)Bevacizumab plus irinotecan, fluorouracil, and leu-covorin for metastatic colorectal cancer. N Engl J Med350: 23352342.

    Innocenti, F., Undevia, S.D., Iyer, L., Chen, P.X.,Das, S., Kocherginsky, M. et al. (2004) Geneticvariants in the Udp-glucuronosyltransferase 1a1 genepredict the risk of severe neutropenia of irinotecan.J Clin Oncol 22: 13821388.

    Innocenti, F., Kroetz, D.L., Schuetz, E., Dolan, M.E.,Ramirez, J., Relling, M. et al. (2009) Comprehensivepharmacogenetic analysis of irinotecan neutropeniaand pharmacokinetics. J Clin Oncol 27: 26042614.

    Ippoliti, C. (1998) Antidiarrheal agents for themanagement of treatment-related diarrhea in cancerpatients. Am J Health Syst Pharm 55: 15731580.

    Iyer, L., Das, S., Janisch, L., Wen, M., Ramirez, J.,Karrison, T. et al. (2002) Ugt1a1*28 Polymorphism asa determinant of irinotecan disposition and toxicity.Pharmacogenomics J 2: 4347.

    Jordan, K., Kellner, O., Kegel, T., Schmoll, H.J. andGrothey, A. (2004) Phase II trial of capecitabine/irinotecan and capecitabine/oxaliplatin in advancedgastrointestinal cancers. Clin Colorectal Cancer4: 4650.

    Karthaus, M., Ballo, H., Abenhardt, W., Steinmetz,T., Geer, T., Schimke, J. et al. (2005) Prospective,double-blind, placebo-controlled, multicenter,randomized phase III study with orally administeredbudesonide for prevention of irinotecan (Cpt-11)-induced diarrhea in patients with advanced colorectalcancer. Oncology 68: 326332.

    Keefe, D.M. (2007) Intestinal mucositis:mechanisms and management. Curr Opin Oncol19: 323327.

    Keefe, D. and Anthony, L. (2008) Tyrosine kinaseinhibitors and gut toxicity: a new era in supportivecare. Curr Opin Support Palliat Care 2: 1921.

    Keefe, D.M. and Gibson, R.J. (2007) Mucosal injuryfrom targeted anti-cancer therapy. Support Care Cancer15: 483490.

    Keefe, D.M., Brealey, J., Goland, G.J. and Cummins,A.G. (2000) Chemotherapy for cancer causesapoptosis that precedes hypoplasia in crypts of thesmall intestine in humans. Gut 47: 632637.

    Keefe, D.M., Gibson, R.J. and Hauer-Jensen, M.(2004) Gastrointestinal mucositis. Semin Oncol Nurs20: 3847.

    Kehrer, D.F., Sparreboom, A., Verweij, J., De Bruijn,P., Nierop, C.A., Van De Schraaf, J. et al. (2001)Modulation of irinotecan-induced diarrhea bycotreatment with neomycin in cancer patients.Clin Cancer Res 7: 11361141.

    Kim, T.W. and Innocenti, F. (2007) Insights,challenges, and future directions in irinogenetics.Ther Drug Monit 29: 265270.

    Kozelsky, T.F., Meyers, G.E., Sloan, J.A., Shanahan,T.G., Dick, S.J., Moore, R.L. et al. (2003) Phase IIIdouble-blind study of glutamine versus placebo for theprevention of acute diarrhea in patients receiving pelvicradiation therapy. J Clin Oncol 21: 16691674.

    Kweekel, D.M., Gelderblom, H., Van Der Straaten,T., Antonini, N.F., Punt, C.J. and Guchelaar, H.J.(2008) Ugt1a1*28 genotype and irinotecan dosage inpatients with metastatic colorectal cancer: a dutchcolorectal cancer group study. Br J Cancer99: 275282.

    Lenfers, B.H., Loeffler, T.M., Droege, C.M. andHausamen, T.U. (1999) Substantial activity ofbudesonide in patients with irinotecan (cpt-11) and5-fluorouracil induced diarrhea and failure ofloperamide treatment. Ann Oncol 10: 12511253.

    Li, Y., Ping, X., Yu, B., Liu, F., Ni, X. and Li, J. (2009)Clinical trial: prophylactic intravenous alanyl-glutamine reduces the severity of gastrointestinal tox-icity induced by chemotherapy: a randomised crossovertrial. Aliment Pharmacol Ther 30(5): 452458.

    Liu, C.Y., Chen, P.M., Chiou, T.J., Liu, J.H., Lin,J.K., Lin, T.C. et al. (2008) Ugt1a1*28 polymorphismpredicts irinotecan-induced severe toxicities withoutaffecting treatment outcome and survival in patientswith metastatic colorectal carcinoma. Cancer112: 19321940.

    Llovet, J.M., Ricci, S., Mazzaferro, V., Hilgard, P.,Gane, E., Blanc, J.F. et al. (2008) Sorafenib inadvanced hepatocellular carcinoma. N Engl J Med359: 378390.

    Maroun, J.A., Anthony, L.B., Blais, N., Burkes, R.,Dowden, S.D., Dranitsaris, G. et al. (2007) Preventionand management of chemotherapy-induced diarrheain patients with colorectal cancer: a consensusstatement by the canadian working group on

    A Stein, W Voigt et al.

    http://tam.sagepub.com 61

  • chemotherapy-induced diarrhea. Curr Oncol14: 1320.

    Martenson, J.A., Halyard, M.Y., Sloan, J.A., Proulx,G.M., Miller, R.C., Deming, R.L. et al. (2008) PhaseIII, double-blind study of depot octreotide versusplacebo in the prevention of acute diarrhea in patientsreceiving pelvic radiation therapy: results of NorthCentral Cancer Treatment Group N00ca. J Clin Oncol26: 52485253.

    Mattison, L.K., Fourie, J., Desmond, R.A., Modak,A., Saif, M.W. and Diasio, R.B. (2006a) Increasedprevalence of dihydropyrimidine dehydrogenasedeficiency in African-Americans compared withCaucasians. Clin Cancer Res 12: 54915495.

    Mattison, L.K., Fourie, J., Hirao, Y., Koga, T.,Desmond, R.A., King, J.R. et al. (2006b) Theuracil breath test in the assessment of dihydropyrimi-dine dehydrogenase activity: pharmacokineticrelationship between expired 13CO2 andplasma [2-13c]dihydrouracil. Clin Cancer Res12: 549555.

    McCollum, A.D., Catalano, P.J., Haller, D.G., Mayer,R.J., Macdonald, J.S., Benson 3rd, A.B. et al. (2002)Outcomes and toxicity in African-American andCaucasian patients in a randomized adjuvantchemotherapy trial for colon cancer. J Natl Cancer Inst94: 11601167.

    Meyerhardt, J.A., Tepper, J.E., Niedzwiecki, D.,Hollis, D.R., McCollum, A.D., Brady, D. et al. (2004)Impact of body mass index on outcomes andtreatment-related toxicity in patients with stage II andIII rectal cancer: findings from Intergroup Trial 0114.J Clin Oncol 22: 648657.

    Michael, M., Brittain, M., Nagai, J., Feld, R., Hedley,D., Oza, A. et al. (2004) Phase II study ofactivated charcoal to prevent irinotecan-induceddiarrhea. J Clin Oncol 22: 44104417.

    Motzer, R.J., Escudier, B., Oudard, S., Hutson, T.E.,Porta, C., Bracarda, S. et al. (2008) Efficacy ofeverolimus in advanced renal cell carcinoma: adouble-blind, randomised, placebo-controlled phaseIII trial. Lancet 372: 449456.

    Motzer, R.J., Hutson, T.E., Tomczak, P., Michaelson,M.D., Bukowski, R.M., Oudard, S. et al. (2009)Overall survival and updated results for sunitinibcompared with interferon alfa in patients withmetastatic renal cell carcinoma. J Clin Oncol27: 35843590.

    Osterlund, P., Ruotsalainen, T., Korpela, R., Saxelin,M., Ollus, A., Valta, P. et al. (2007) Lactobacillussupplementation for diarrhoea related tochemotherapy of colorectal cancer: a randomisedstudy. Br J Cancer 97: 10281034.

    Pan, C.X., Loehrer, P., Seitz, D., Helft, P., Juliar, B.,Ansari, R. et al. (2005) A phase II trial ofirinotecan, 5-fluorouracil and leucovorincombined with celecoxib and glutamine as first-linetherapy for advanced colorectal cancer. Oncology69: 6370.

    Pytlik, R., Benes, P., Patorkova, M., Chocenska, E.,Gregora, E., Prochazka, B. et al. (2002) Standardizedparenteral alanyl-glutamine dipeptide supplementationis not beneficial in autologous transplant patients: arandomized, double-blind, placebo controlled study.Bone Marrow Transplant 30: 953961.

    Reardon, D.A., Quinn, J.A., Vredenburgh, J., Rich,J.N., Gururangan, S., Badruddoja, M. et al. (2005)Phase II trial of irinotecan plus celecoxib in adults withrecurrent malignant glioma. Cancer 103: 329338.

    Rosenoff, S. (2004a) Resolution of refractorychemotherapy-induced diarrhea (CID) with octreotidelong-acting formulation in cancer patients: 11 casestudies. Support Care Cancer 12: 561570.

    Rosenoff, S.H. (2004b) Octreotide Lar resolves severechemotherapy-induced diarrhoea (CID) and allowscontinuation of full-dose therapy. Eur J Cancer Care(Engl) 13: 380383.

    Rosenoff, S.H., Gabrail, N.Y., Conklin, R., Hohneker,J.A., Berg, W.J., Warsi, G. et al. (2006) A multicenter,randomized trial of long-acting octreotide for theoptimum prevention of chemotherapy-induceddiarrhea: results of the stop trial. J Support Oncol4: 289294.

    Saliba, F., Hagipantelli, R., Misset, J.L., Bastian, G.,Vassal, G., Bonnay, M. et al. (1998) Pathophysiologyand therapy of irinotecan-induced delayed-onset diar-rhea in patients with advanced colorectal cancer: aprospective assessment. J Clin Oncol 16: 27452751.

    Saltz, L.B., Cox, J.V., Blanke, C., Rosen, L.S.,Fehrenbacher, L., Moore, M.J. et al. (2000) Irinotecanplus fluorouracil and leucovorin for metastatic color-ectal cancer. Irinotecan Study Group. N Engl J Med343: 905914.

    Saltz, L.B., Niedzwiecki, D., Hollis, D., Goldberg,R.M., Hantel, A., Thomas, J.P. et al. (2007) Irinotecanfluorouracil plus leucovorin is not superior to fluor-ouracil plus leucovorin alone as adjuvant treatment forstage III colon cancer: results of Calgb 89803. J ClinOncol 25: 34563461.

    Schmittel, A., Jahnke, K., Thiel, E. and Keilholz, U.(2004) Neomycin as secondary prophylaxis foririnotecan-induced diarrhea. Ann Oncol 15: 1296.

    Sergio, G.C., Felix, G.M. and Luis, J.V. (2008)Activated charcoal to prevent irinotecan-induceddiarrhea in children. Pediatr Blood Cancer 51: 4952.

    Siber, G.R., Mayer, R.J. and Levin, M.J. (1980)Increased gastrointestinal absorption of large mole-cules in patients after 5-fluorouracil therapy for meta-static colon carcinoma. Cancer Res 40: 34303436.

    Stringer, A.M., Gibson, R.J., Logan, R.M., Bowen,J.M., Yeoh, A.S.J., Burns, J. et al. (2007)Chemotherapy-induced diarrhea is associated withchanges in the luminal environment in the da rat.Experimental Biology and Medicine 232: 96106.

    Stringer, A.M., Gibson, R.J., Logan, R.M.,Bowen, J.M., Yeoh, A.S. and Keefe, D.M. (2008)

    Therapeutic Advances in Medical Oncology 2 (1)

    62 http://tam.sagepub.com

  • Faecal microflora and beta-glucuronidase expressionare altered in an irinotecan-induced diarrhea model inrats. Cancer Biol Ther 7: 19191925.

    Stringer, A.M., Gibson, R.J., Logan, R.M., Bowen,J.M., Yeoh, A.S., Laurence, J. et al. (2009)Irinotecan-induced mucositis is associated withchanges in intestinal mucins. Cancer ChemotherPharmacol 64: 123132.

    Takasuna, K., Kasai, Y., Kitano, Y., Mori, K.,Kakihata, K., Hirohashi, M. et al. (1995) [Study onthe mechanisms of diarrhea induced by a new antic-ancer camptothecin derivative, irinotecan hydrochlor-ide (Cpt-11), in Rats]. Nippon Yakurigaku Zasshi105: 447460.

    Takasuna, K., Hagiwara, T., Hirohashi, M., Kato, M.,Nomura, M., Nagai, E. et al. (1998) Inhibition ofintestinal microflora beta-glucuronidase modifies thedistribution of the active metabolite of the antitumoragent, irinotecan hydrochloride (Cpt-11) in rats.Cancer Chemother Pharmacol 42: 280286.

    Threadgill, D.W., Dlugosz, A.A., Hansen, L.A.,Tennenbaum, T., Lichti, U., Yee, D. et al. (1995)Targeted disruption of mouse EGF receptor: effect ofgenetic background on mutant phenotype. Science269: 230234.

    Trifan, O.C., Durham, W.F., Salazar, V.S., Horton, J.,Levine, B.D., Zweifel, B.S. et al. (2002)Cyclooxygenase-2 inhibition with celecoxib enhancesantitumor efficacy and reduces diarrhea side effect ofCpt-11. Cancer Res 62: 57785784.

    Van Cutsem, E., Peeters, M., Siena, S., Humblet, Y.,Hendlisz, A., Neyns, B. et al. (2007) Open-label phaseIII trial of panitumumab plus best supportive carecompared with best supportive care alone in patientswith chemotherapy-refractory metastatic colorectalcancer. J Clin Oncol 25: 16581664.

    Van Cutsem, E., Kohne, C.H., Hitre, E., Zaluski, J.,Chang Chien, C.R., Makhson, A. et al. (2009)Cetuximab and chemotherapy as initial treatment formetastatic colorectal cancer. N Engl J Med360: 14081417.

    Vincenzi, B., Schiavon, G., Pantano, F., Santini, D.and Tonini, G. (2008) Predictive factors forchemotherapy-related toxic effects in patients withcolorectal cancer. Nat Clin Pract Oncol 5: 455465.

    Voigt, W., Matsui, S., Yin, M.B., Burhans, W.C.,Minderman, H. and Rustum, Y.M. (1998)Topoisomerase-I inhibitor Sn-38 can induce DNAdamage and chromosomal aberrations independentfrom DNA synthesis. Anticancer Res 18: 34993505.

    Von Bultzingslowen, I., Adlerberth, I., Wold, A.E.,Dahlen, G. and Jontell, M. (2003) Oral and intestinal

    microflora in 5-fluorouracil treated rats, translocationto cervical and mesenteric lymph nodes and effects ofprobiotic bacteria. Oral Microbiol Immunol18: 278284.

    Wadler, S., Haynes, H. and Wiernik, P.H. (1995)Phase I trial of the somatostatin analog octreotideacetate in the treatment of fluoropyrimidine-induceddiarrhea. J Clin Oncol 13: 222226.

    Wadler, S., Benson 3rd, A.B., Engelking, C., Catalano,R., Field, M., Kornblau, S.M. et al. (1998)Recommended guidelines for the treatment ofchemotherapy-induced diarrhea. J Clin Oncol16: 31693178.

    Wasserman, E., Hidalgo, M., Hornedo, J. and Cortes-Funes, H. (1997b) Octreotide (Sms 201-995) forhematopoietic support-dependent high-dose chemo-therapy (Hsd-Hdc)-related diarrhoea: dose findingstudy and evaluation of efficacy. Bone MarrowTransplant 20: 711714.

    Wasserman, E., Myara, A., Lokiec, F., Goldwasser, F.,Trivin, F., Mahjoubi, M. et al. (1997a) Severe Cpt-11toxicity in patients with Gilberts syndrome: two casereports. Ann Oncol 8: 10491051.

    Xue, H., Sawyer, M.B., Field, C.J., Dieleman, L.A.,Murray, D. and Baracos, V.E. (2008) Bolus oralglutamine protects rats against Cpt-11-induceddiarrhea and differentially activates cytoprotectivemechanisms in host intestine but not tumor. J Nutr138: 740746.

    Yen, J.L. and McLeod, H.L. (2007) Should DPDanalysis be required prior to prescribingfluoropyrimidines? Eur J Cancer 43: 10111016.

    Zachariah, B., James, J., Gwede, C.X., Ajani, J., Chin,L., Donath, D. et al. (2007) RTOG 0315: arandomized, double-blind, placebo-controlledphase III study to determine the efficacy ofoctreotide acetate in preventing or reducing theseverity of chemoradiation-induced diarrhea inpatients with anal or rectal cancer. J Clin Oncol2007 ASCO Annual Meeting Proceedings Part I25: 4032.

    Zalcberg, J., Kerr, D., Seymour, L. and Palmer, M.(1998) Haematological and non-haematologicaltoxicity after 5-fluorouracil and leucovorin in patientswith advanced colorectal cancer is significantly asso-ciated with gender, increasing age and cycle number.Tomudex International Study Group. Eur J Cancer34: 18711875.

    Zidan, J., Haim, N., Beny, A., Stein, M., Gez, E. andKuten, A. (2001) Octreotide in the treatment of severechemotherapy-induced diarrhea. Ann Oncol12: 227229.

    Visit SAGE journals onlinehttp://tam.sagepub.com

    A Stein, W Voigt et al.

    http://tam.sagepub.com 63